Sie sind auf Seite 1von 11

Hindawi Publishing Corporation

BioMed Research International


Volume 2013, Article ID 297501, 11 pages
http://dx.doi.org/10.1155/2013/297501

Review Article
Genetic and Functional Profiling of Crohn’s Disease: Autophagy
Mechanism and Susceptibility to Infectious Diseases

Annalisa Marcuzzi,1 Anna Monica Bianco,1 Martina Girardelli,1 Alberto Tommasini,1


Stefano Martelossi,1 Lorenzo Monasta,1 and Sergio Crovella1,2
1
Institute for Maternal and Child Health—IRCCS “Burlo Garofolo” of Trieste, Via dell’Istria 65/1, 34137 Trieste, Italy
2
University of Trieste, 34127 Trieste, Italy

Correspondence should be addressed to Annalisa Marcuzzi; marcuzzi@burlo.trieste.it

Received 27 February 2013; Accepted 20 March 2013

Academic Editor: Enrique Medina-Acosta

Copyright © 2013 Annalisa Marcuzzi et al. This is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.

Crohn’s disease is a complex disease in which genome, microbiome, and environment interact to produce the immunological
background of the disease. Disease in childhood is more extensive and characterized by a rapid progression, leading to severe
repercussions in the course of the disorder. Several genetic variations have been associated with an increased risk of developing the
disease and most of these are also implicated in other autoimmune disorders. The gut has many tiers of defense against incursion by
luminal microbes, including the epithelial barrier and the innate and adaptive immune responses. Moreover, recent evidence shows
that bacterial and viral infections, as well as inflammasome genes and genes involved in the autophagy process, are implicated in
Crohn’s disease pathogenesis. The aim of this review is to establish how much the diagnostic system can improve, thus increasing the
success of Crohn’s disease diagnosis. The major expectation for the near future is to be able to anticipate the possible consequences
of the disease already in childhood, thus preventing associated complications, and to choose the best treatment for each patient.

1. Introduction of steroids to control the first flare [8]. However, as the


median age at diagnosis is 27 years, patients may live with
Crohn’s disease (CD) is a chronic form of inflammatory bowel CD for more than 50 years in the Western world, where life
disease (IBD) that can affect any part of the gastrointestinal expectancy of patients exceeds 70 years.
tract, from the mouth to the anus. However, it most com- The age of onset is frequently in the second decade
monly affects the colon and terminal ileum [1] with up to of life, and most patients progress to a relapsing disease
75% of patients having ileal disease with or without colonic characterized by abdominal pain, bloody diarrhea, vomit,
involvement [2]. It is a debilitating disorder with an overall and weight loss.
prevalence of 0.5%–1% of the general population [3, 4]. CD Although CD normally manifests in adulthood, it can
differs from other types of IBDs because in patients with CD be present in childhood before the age of 2 years [9].
the inflammation is often continuous and with involvement The early onset Crohn’s disease (EOCD) is typically more
of the mucosa [5]. extensive (beyond the colon and/or oral or perianal dis-
Complications are common but not a constant: disease ease) and characterized by rapid progression, leading to
progression is marked by severe colitis, strictures and peri- severe repercussions in disease development [10]. Diagnosis
anal fistulas, typically requiring surgery [6, 7]. is particularly challenging in children in which presenting
Beaugerie et al. [8] recently reported three factors that at symptoms may vary widely and may only consist of subtle
the time of the diagnosis increase the chance of developing a extraintestinal manifestations [11]. This often leads to a typical
disabling disease in the following five years: (A) age <40 years, delay in the diagnosis of pediatric IBD, ranging from 4 weeks
(B) presence of perianal lesions, and (C) the requirement in severe colitis to 6-7 months in milder disease. Reducing
2 BioMed Research International

this diagnostic delay is important, since a long period of


unmanaged symptoms can significantly impact on growth
Genetic
and early treatment is essential to preserve long-term quality susceptibility
of life [12, 13]. Thus, a sensitive yet noninvasive tool for
the identification of patients at high risk of IBD, and there-
fore warranting endoscopic evaluation, would be a valuable
diagnostic aid. There are specific clinical, therapeutic, and
Crohn’s
psychosocial issues specific to children with IBD that must disease
be considered to ensure prompt diagnosis and appropriate
medical management.
The etiopathogenesis of CD is unclear. It remains to Infectious Immune
be determined whether this disease represents an abnormal disease response
response to normal antigenic stimuli or an appropriate
response to persistently abnormal stimuli [14, 15]. A better
understanding of the origin of the disease and the mecha-
Figure 1: Crohn’s disease is a heterogeneous disorder of multifacto-
nisms of action is necessary to improve the prognosis of CD. rial etiology in which genetic, environmental, and microbial factors,
CD is a complex disorder resulting from the interaction of together with the immunological response, interact to produce the
genetic environmental and microbial factors. Given the diffi- disease.
cult genotype-phenotype correlation and given the heteroge-
neous genetics, the different interactions among predisposing
factors, and not only the number of genes involved, should the immune response against the luminal flora in a genetically
be considered in order to understand the mechanisms of this susceptible host [21] (Figure 1).
disease. Many previous and ongoing studies have sought to It is commonly assumed that CD is a heterogeneous disor-
identify genes and especially disease-causing variants such as der of multifactorial etiology in which genome, microbiome
risk factors for the disease. Identification and characterization (hereditability), and environment interact to produce the
of disease-causing variants represents one of the biggest chal- immunological background of the disease. It is probable that
lenges of genetics within the etiopathogenetic study of CD. patients have a genetic predisposition for the development
Genomewide association studies already identified several of the disease coupled with immunoregulation disturbances
distinct genetic polymorphisms associated with Crohn’s dis- [22].
ease. In European individuals, NOD2 gene polymorphisms
confer by far the greatest risk for the disease. Other variations
2.1. Genetic Susceptibility: Greater Weighing Factor in Early
in ATG16L1, IRGM, and IL23R genes were reported to be
Onset Crohn Disease. The epidemiologic evidence of the
highly associated with CD.
role of genetic factors in the pathogenesis of the dis-
The genetic background has certainly a predisposing
ease came from studies demonstrating higher rates of CD
role, but several alternative explanations are possible, mostly
among individuals of Caucasian and Jewish ethnicity, familial
related to lifestyle. The importance of environmental factors is
aggregation of CD, and higher concordance rates of both
shown by an increase in the incidence rate of diseases in eth-
twins developing CD in monozygotic compared to dizygotic
nic groups previously less affected, as Hispanic, Asians, and
twins. Due to the complexity of the disease, the search for
immigrants that had moved from regions of low incidence
specific CD susceptibility genes has been very difficult so
into areas where the incidence of the disease is higher [16].
far. Despite the large number of genomewide associations
Observation of Crohn’s patients and animal models sug- (GWAS) established to date, most complex diseases (not
gests a role of bacteria in the disorder [17]. Among the most monogenic) have only managed to explain some additional
important bacteria that can adhere and invade the mucosa is percentage of the hereditability estimates. The source of this
Escherichia coli [18]. The onset of the disease is quite common missing hereditability is the subject of much debate with
after gastrointestinal infections and people suffering from this various explanations: overestimates of original heritability
disorder have, generally, higher concentrations of mucosal statistics, underpowered GWAS studies to detect common
bacteria if compared to healthy subjects [19]. variants, poorly investigated epistasis and gene-environment
A detailed picture of how genes work together and interactions, and rare genetic variants [23]. In the attempt
interact with environmental and microbial factors may better to explain some of this missing hereditability, researchers
explain individual differences in CD susceptibility. have adopted several complementary strategies. Combined
genotypes, “private genes,” and epigenetic markers may
account for this missing hereditability: monogenic immunity
2. Etiology of CD disorders are increasingly diagnosed in patients with EOCD
[24]. Advances in bioinformatics have now made it possible
The complex pathophysiology of CD has, for a long time, been to perform GWAS using copy number variation probes. By
an enigma [20]. several GWAS and meta-analysis studies many genes have
Although the precise etiology of CD remains elusive, been associated with CD: more than 90 distinct genomic loci
epidemiological data conclusively indicate a deregulation of have been found to be associated with an increased risk of
BioMed Research International 3

Genetics
susceptibility loci Autophagy
NOD2, ATG16, IRGM,
LRRK2

New loci CD associated [23] Other Th17 pathway


ZMIZ1, FUT2, CPEB4, IL23R, IL12B, STAT3,
5q31, IKZF1, AK2, TYK2
ZPBP2TNFS18-4, FASLG,,
ORMAD2 ADAM30, IL6ST,
Early-onset associated loci [30] ILL31R, CREB5, AZF1,
RIPK2, RASGRP1, SPRED1,
LGALS9, NOS2, IFNGR2-1, Immune-mediated
CREM, C11ORF30, PTPN22, CCR6, IL2RA,
PTGER4, ORMDL3, IL18RAP,
IBD associated loci GSMDL 16p11.2, ERAP2,
ITLN1,
CCL2/CCL7,
TNFSF11, BACH2,
TAGAP, VAMP,
TNFSF15, ICOSLG

Figure 2: More than 90 distinct genomic susceptibility loci have been found to be associated with an increased risk of developing CD. The
genes variants relate largely to the innate immunity genes, in particular to the disruption of the innate and adaptative arms of the immune
systems, to the process of autophagy, to the epithelial barrier function, and to the activation of the endoplasmic reticulum stress response.

developing CD. Genes with replicated evidence for strong recent GWAS focused on a pediatric cohort highlighting the
association suggest that these variants relate largely to the implication of novel pathways and interaction between the
innate immunity genes, in particular to the disruption of two different onsets [35, 36].
the innate and adaptive arms of the immune system, to the Moreover, environmental factors such as smoking are less
process of autophagy, to the epithelial barrier function, and likely to be exerting an influence on the disease in pediatric
to the activation of the endoplasmic reticulum stress response cohorts [37].
[25–27] (Figure 2).
Most loci are in relation with the CD phenotype and 2.2. Gastrointestinal Microbiota: Host Genome-Microbe Inter-
many loci are implicated in other immune-mediated dis- actions in Crohn’s Pathogenesis. Considering epidemiolog-
orders, most notably with ankylosing spondylitis, erythema ical, genetic and immunological data, it is probable that
nodosum, and psoriasis [28]. Several genes are involved in patients have a genetic predisposition for the development of
primary immunodeficiencies, characterized by a dysfunc- the disease coupled with disturbances in both immunoregu-
tional immune system resulting in severe infections [26, 29, lation, and intestinal microbiota. The disease can be triggered
30]. In the last years defective processing of intracellular by any of a number of different factors and sustained by
bacteria has become a central theme. A considerable overlap an abnormal immune response to these factors. Rather,
has also been observed between susceptibility loci for CD and the intensive interaction between intestinal epithelial cells
susceptibility for infectious diseases [27]. and immune competent cells is critical to maintain and
Genetic susceptibility is thought to play a more important perpetuate the chronic inflammatory process characteristic
role in the etiology of early rather than late onset CD [31]. of CD [22]. The genetic and pathological complexity of CD
This is supported by a higher rate of positive family history of is particularly well suited for testing whether interactively
CD in patients with a younger age at diagnosis with respect redefining disease diagnoses can enhance the value of genetic
to patients with older age at diagnosis, suggesting that an and pathogenetic studies. Precision in the characterization of
earlier presentation may be due to a higher burden of disease- the disease would make defining the impact of host-gene-
causing mutations in the genomes of these affected children microbial interactions on the disease process more robust.
compared to those in whom disease manifests later in life The human body is inhabited by a vast number of
[32]. EOCD presents a more aggressive phenotype; in fact bacteria, archaea, viruses, and unicellular eukaryotes. The
earlier age at diagnosis is associated with a greater need for microbiota represents a collection of microorganisms that
surgery [33]. EOCD candidate susceptibility genes have been live in peaceful coexistence with their hosts [38]. By far,
identified using linkage analysis and gene sequencing in two the most heavily colonized organ is the gastrointestinal tract
unrelated consanguineous families [34]. Some gene/loci may (GIT) and the colon alone is estimated to contain over
be specific to pediatric-onset CD and that is documented by 70% of all the microbes [39] and represents a major surface
4 BioMed Research International

Genetic
Environmental
susceptibility
factors
Pathological gut
Healthy gut

PAMPs Lumen Defensins


IgA, IgG

Mucus layer

Epithelial barrier
Damaged
epithelial
barrier

Lamina propria
Physiological inflammation

Chronic inflammation
Homeostasis

Commensal Autoinflammatory Mucolytic Harmful


bacteria bacteria bacteria bacteria

Figure 3: The role of microbiota in Crohn’s disease pathogenesis. The interplay between the host microbiota and the environmental factors
in a genetic susceptible host results in a progressive inflammatory damage to the host intestinal mucosa.

for microbial colonization. It is estimated that the human major bacterial phyla: Firmicutes, Bacteroidetes, and to a
microbiota, not homogeneous in the GIT, contains as many as lesser degree Proteobacteria and Actinobacteria [50]. Many
1014 bacterial cells [40, 41] and the number of bacterial species studies have observed imbalances or dysbioses in the GI
present in the human gut is estimated to be 500 to 1000 [42]. microbiomes of CD patients [51, 52]. In CD patients biodiver-
Nevertheless, a recent analysis involving multiple subjects sity is decreased, with a lower proportion of Firmicutes and an
has suggested that the collective human gut microbiota is increase in Gammaproteobacteria [53]. In CD, proportions of
composed of over 35000 bacterial species [43]. the Clostridia are altered: the Roseburia and Faecalibacterium
Colonization of the human gut with microbes begins genera of the Lachnospiraceae and Ruminococcaceae families
immediately at birth; in fact infants are exposed to a complex are decreased, whereas Ruminococcus gnavus increases [54].
microbial population upon the passage through the birth An important concept in the pathogenesis of CD is that
canal [44] and it is known that infants delivered through bacterial and viral interactions occur in a host gene-specific
cesarean section have different microbial compositions com- manner [55, 56]. Surgical diversion of the fecal stream
pared to vaginally delivered infants [45]. It has been shown ameliorates inflammation. In addition, the evaluation of the
that the microbiota of adult monozygotic and dizygotic twins microbial populations in surgically resected tissue samples
was equally similar to that of their siblings, suggesting that of small bowel and colon from CD patients and non-CD
the colonization by the microbiota from a shared mother controls, by rRNA sequence analysis, showed that specific
was more decisive in determining their adult microbiota than flora was not enriched in small bowel or colon from CD
their genetic makeup [46] (Figure 3). patients. However, a subset of CD samples showed alterations
Several studies have shown that host genetics can impact in the representations of the Bacteroides and Firmicutes
the microbial composition of the gut [47, 48]. The central role [43, 57]. Moreover, several studies have shown that the gut
of gut microbiota in the development of mucosal immunity microbiota is altered in IBD patients. For example, biopsy
is not surprising considering that the intestinal mucosa samples from CD patients were used to prepare bacterial
represents the largest surface area in contact with the anti- DNA which was amplified using universal bacterial 16S rRNA
gens of the external environment called PAMPs (pathogen- primers [58], and a significant increase in Proteobacteria and
associated molecular patterns). Additionally, the dense car- Bacteroidetes was found in CD patients compared to controls,
pet of the gut microbiota overlying the mucosa normally with a decrease in Clostridia. Metagenomic approaches were
accounts for the largest proportion of the antigens presented used to analyze fecal samples from Crohn’s patients and
to the resident immune cells and those stimulating the pattern healthy donors and revealed reduced complexity of the
recognition receptors such as the NOD-like receptors (NLRs) Firmicutes in affected individuals [59]. Finally, the evidence
of the intestinal epithelial cells [49]. The GI (gastrointestinal that intestinal bacteria play an important role in CD patients
gut) microbiome of healthy humans is dominated by four is that antibiotics help some patients and can ameliorate
BioMed Research International 5

IL-17A
IL-17F
IL-21
IL-22 IFN-𝛾
IL-26 TNF-𝛼
IL-6 Th17 IL-6
TNF-𝛼

Th1 IL-10
TGF-𝛽

Proinflammatory cytokines Treg

Anti-inflammatory cytokines

Figure 4: Up- and Downregulation of the proinflammatory cytokines evidenced in the immune system dysregulation of CD patients.

disease activity. Moreover metronidazole is an important cells. The discovery that Th17 cells, which express the IL-
therapeutic agent for certain complications of CD such as 23 receptor (IL-23R), play a role in CD pathogenesis was
fistulising disease. Viral infection is required to generate the supported by recent GWAS studies demonstrating that IL-
Paneth cell defect found in ATG16L1 mice [60], suggesting 23R and other genes involved in the differentiation of Th17
that in addition to human bacterial microbiota, viral or fungal cells are susceptibility genes.
commensals may play a role in CD pathogenesis. To confirm the link between immune response and
genetic susceptibility in the pathogenesis of CD there are
several recent lines of evidence that the key role is played
2.3. Immunological Response in CD
by autophagy that includes the antigen presentation and the
2.3.1. Th1 and Th17 Implicated in CD Pathogenesis. Although production of proinflammatory cytokines. The relationship
the exact CD etiology is still not completely understood, between autophagy and microbes, indeed, has remained ill-
several studies indicate that its pathogenesis is characterized defined until a recent convergence of studies showing that
by an exaggerated immune response in genetically susceptible autophagy is an innate immune defense against bacteria, pro-
individuals. tozoa, and viral pathogens [64]. It is commonly assumed that
CD patients suffer from marked immune system dereg- the role of autophagy in addition to eliminating intracellular
ulation. The inflammation seen in these patients is charac- pathogens [65] contributes to MHC II restricted endogenous
terized by pronounced Th1 and Th17 responses [61] involving antigen presentation. It is an effector of Th1/Th2 polarization,
upregulation of proinflammatory cytokines IL-1, IL-6, IL-12, affects B and T cell homeostasis and repertoire selection,
TNF-𝛼, IFN-𝛾, IL-23, and IL-17 and downregulation of IL- 10, delivers cytosolic PAMP or danger associated molecular
but it is not clear whether this is a cause or a consequence of patterns to endosomal toll-like receptors (TLR), and acts
the disease [62] (Figure 4). as an innate immunity effector downstream of TLR [66].
Th1 cells are commonly assumed to be associated with CD Polymorphisms in autophagy genes result in deregulation of
development and produce IFN- 𝛾, and their primary role is these processes and affect gut homeostasis: genetic variants of
the protection against intracellular microbes. IFN-𝛾 secreting autophagy genes have been linked to CD.
lamina propria lymphocytes are abundant in the mucosa of
CD patients: this condition is marked at CD onset (mucosal
T cells appear to mount a typical Th1 response that resembles 2.3.2. The Role of the NLRP3 Inflammasome in the Patho-
an acute infectious process) and disappears in late CD. genesis of CD. Inflammasomes are cytoplasmic multipro-
Recently, several studies showed the pivotal role of the tein complexes that function as sensors of endogenous
imbalance of regulatory T cells (Treg) and Th17 in CD. Treg or exogenous PAMPs. They are composed of one of sev-
cells are important for the control of the immune response to eral nucleotide-binding oligomerization-domain protein-like
self-antigens preventing autoimmunity and maintaining self- receptors (NLRs), including NLRP1, NLRP3, NLRP6, and
tolerance [63]. In contrast, IL-17 producing Th17 cells were NLRPC4. Upon sensing the relevant signal, they assemble,
recognized as a novel group of T cells which play a major typically together with an adaptor protein, an apoptosis-
role in autoimmunity. The gastrointestinal immune system associated speck-like protein (ASC) or a caspase activating
has to maintain both a state of tolerance toward intestinal and recruitment domain 8 (CARD8), into a multiprotein
antigens and the ability to combat pathogens. In CD this complex that governs caspase-1 activation and subsequent
balance is lost and the effects of proinflammatory T cells cleavage of effector proinflammatory cytokines including
outnumber the tolerizing, anti-inflammatory effects of Treg pro-IL-1𝛽 and pro-IL-18.
6 BioMed Research International

Recently several studies highlighted with particular only within the gut, probably owing to the high microbial load
emphasis the relevance and the role of NLRP3 (previously in this tissue.
known as CIAS1 and NLRP3) in the pathogenesis of CD Nucleotide-binding-oligomerization-domain- (NOD-)
[67, 68]. like receptors (NLRs) represent ancient sentinels of the host
There is evidence suggesting that NLRP3 is able to innate immune system, and genetic variants in NLR genes
respond to a variety of signals: adenosine triphosphate are associated with complex chronic inflammatory barrier
(ATP), nigericin, maitotoxin, Staphylococcus aureus and Lis- diseases [85]. The NOD2 gene is an intracellular sensor for
teria monocytogenes [69], and RNA and uric acid crystals the bacterial cell wall component muramyl-dipeptide, and
(monosodium urate and calcium pyrophosphate dehydrate) loss-of-function variants in the human NOD2 gene have
released from dying cells [70, 71]. been associated with an increased susceptibility for CD
Literature data showed that the proinflammatory com- [86, 87] in Caucasian populations of European ancestry [88],
pound muramyl-dipeptide, the minimal bioactive peptido- and particularly for ileal disease [89], and were found to be
glycan motif common to all bacteria, was an activator of the an important regulator of the commensal gut microbiota in
NLRP3 inflammasome, which suggested a very interesting mice [90]. NOD2 recognizes components of the bacterial cell
connection between NOD2 and NALP3 [72]. wall and elicits an NF-𝜅B response and mediates the release
On the other hand, Kanneganti et al. suggested that of defensins, which are antimicrobial peptides. Evidence
bacterial RNA and small antiviral compounds are the specific from MDP stimulation of NOD2-activated autophagy shows
ligands of NLRP3 rather than MDP [70]. a link between genetic risk loci and highlights the importance
Anyway, NLRP3 plays a pivotal role in the inflammation of defining disease associated pathways and the potential of
regulating the activation of the caspase-1 and processing of new roles for known genes [78]. Epithelial cells and dendritic
IL-1𝛽, two key mediators involved in the pathogenesis of the cells containing Crohn’s-disease-associated ATG16L1 and
more common inflammatory disorders [73]. NOD2 variants show defects in antibacterial autophagy
More recently, in an increasingly complicated picture, [79, 91]. In dendritic cells, these defects are associated with
Elinav et al. described a novel regulatory sensing system in an impaired ability to present exogenous antigens to CD4+T
the colon, dependent on the NLRP6 inflammasome [74], and cells [78]. A discussed model of Crohn’s disease is the one in
von Kampen et al. showed that CARD8 negatively regulates which individuals are genetically susceptible to a pathogen
NOD-2 mediated signaling [75]. These current data further that triggers a compensatory and harmful immune response.
underline the link between the different components of the Antibacterial autophagy, through ATG16L1, NOD2, and
CD etiopathogenesis that are strongly correlated. potentially other genes (IRGM), is consistent with this
model. However, one of the most important experimental
supports for this model comes from an unrelated study using
3. Autophagy in CD Citrobacter rodentium to induce intestinal inflammation
in NOD2 –/– mice [92]. These results illustrate a close
Genomewide association studies and genetic analyses have relationship between NOD2, ATG16L1, and autophagy,
emphasized the involvement of autophagy processes in the affecting intracellular processing and communication
pathogenesis of inflammatory bowel diseases implicating with the adaptive immune system suggesting that genetic
three component genes in CD pathogenesis: ATG16L1 [76], polymorphisms may affect both pathways concomitantly.
IRGM [77], and NOD2 [78, 79]. These genes encode proteins IRGM belongs to the p47 Immunity-Related GTPase
critical for autophagy, a process that mediates degradation (IRG) family and is linked to CD by GWAS as a protein
of intracellular proteins via vesicle-mediated delivery to the that is implicated in the autophagy mechanism [93]. The
lysosome [80, 81]. Autophagy is involved in intracellular analysis of the interactions between 44 autophagy-associated
homeostasis, contributing to the degradation and recycling human proteins and 83 viral proteins belonging to different
of cytosolic contents and organelles, as well as to resistance RNA virus families revealed that IRGM was the autophagy-
against infection and the removal of intracellular microbes. It associated protein most targeted by these viruses. IRGM can
is a major degradative pathway of the cell with several critical interact with 12 viral proteins belonging to different viruses,
functions in innate and adaptive immunity [82] (Figure 5). such as HCV and HIV-1 [94, 95]. A recent study suggests
The ATG16L1 deficiency mouse showed Paneth cell dys- that a polymorphism in IRGM could affect the binding and
function with aberrant exocytosis, as well as an altered the consequent misregulation by a specific miRNA (miR-196)
transcriptional profile, characterized by increased expression that is highly expressed in the intestinal tissue of patients with
of pro-inflammatory cytokines and lipid metabolism genes Crohn’s disease. The consequence is that the xenophagy flux is
like Paneth cell phenotype of CD patients [83]. Nevertheless not well regulated leading to the accumulation of bacteria in
an important observation derived from the ATG16L1 mouse the lysosomal compartment. This study showed that greater
model was that the murine norovirus infection, as well as IRGM expression leads to both colocalization of adherent
the presence of the commensal bacteria, was required for invasive E. coli (AIEC) with the autophagy machinery and
the generation of these specific Paneth cell abnormalities increased intracellular survival of the bacteria [96]. This and
[84]. ATG16L1 is essential for all forms of autophagy, and the other strains of E. coli are more abundant in the mucosa of
coding mutation T300A is associated with increased risk of CD patients [90]. The ability of IRGM to induce autophagy
CD. Despite its ubiquitous expression, the defects associated and limit the replication of intracellular bacteria has been
with ATG16L1 polymorphisms have so far been described demonstrated with mycobacteria by inducing mitochondrial
BioMed Research International 7

MDP
Bacteria

CARD1 CARD 2 NOD LRR


Plasma NOD 2
membrane

CARD1 CARD 2 NOD LRR


CARD1 CARD 2 NOD LRR
(b)
ATG16 WD repeat ATG16 WD repeat
ATG16L1 ATG12
ATG5

(c) IRGM IIGP Endosome

Amphisome
Autolysosome Phagophore

Autophagosome

Lysosome

Degradation Maturation Closure Elongation Initiation


(a)
Autophagy process

Figure 5: CD pathogenesis and autophagy: susceptible CD genes ATG16L1, NOD2, and IRGM are proteins critical for the autophagy
process. (a) The process of mammalian autophagy is divided into the following principal steps: initiation, elongation, closure, maturation,
and degradation. (b) At the bacterial entry site NOD2 activated by MDP recruit ATG16L1 to the plasma membrane. Follow the assembling
of the ATG5-ATG12 complex, stabilized by ATG16L1, that facilitates the formation of an autophagosome around the invading bacterium. (c)
IRGM, another autophagy-related gene, could be involved in the final steps of the degradation step.

depolarization and can increase ROS production and cell more severe clinical manifestation and complication of the
death [97]. Finally IRGM could regulate inflammation by pediatric disease, that make the intestinal mucosal extremely
either regulating intracellular pathogens or cellular home- thin and at risk of perforation.
ostasis much like ATG16L1. Noninvasive tests for CD already exist, including antibod-
These data provide further information and support for ies, imaging-based screens, and fecal biomarkers [98]. The
the hypothesis that microbial/viral interactions with the specificity of existing methods ranges from 89% to 95% for
intestinal mucosa are required for disease generation and CD and other inflammatory bowel diseases. However, these
suggest that combinatorial models for CD pathogenesis are methods are limited to active disease and poorly sensitive
most relevant for the study of human disease pathogenesis. (∼55%). Their outcome can be confounded by other diseases,
further limiting their clinical utility. Recently, high expecta-
4. Concluding Remarks tions are placed in diagnostic studies of the gastrointestinal
microbiota, but further validations will be necessary before
The diagnosis of CD is reached through the results of clinical, this tool is accepted in clinical practice [32, 99].
laboratory, radiographic, endoscopic, and histologic analyses. Research is moving forward in order to identify new and
Radiological and endoscopic techniques are essential for valid biomarkers for the diagnosis of the disease with the aim
the diagnosis of CD since its onset and are useful in assessing of replacing the use of invasive techniques.
the inflammatory status of the intestinal mucosa. However, Currently, only the measurement of fecal calprotectin
endoscopy is an invasive procedure. In children it can be levels has achieved a place in clinical routine practice and is
traumatic and could have critical implications due to the used as a marker for noninvasive determination of intestinal
8 BioMed Research International

inflammation [99, 100]. This protein is an ideal marker In conclusion, it would certainly be useful to be able
because it is not degraded by the human microbiota. Levels of to create a biological algorithm that helps clinicians in the
fecal calprotectin significantly increase in patients with CD, identification and classification of the disease and to deter-
ulcerative colitis, infectious colitis, and, to a lesser extent, mine the pharmacological care. This algorithm could include
in tumors of the colon rectum, but not in patients with not only the known and principal factors predisposing to
functional disorders, as in the case of the irritable bowel the disease, but also the gene-microbiome interaction and
syndrome. In CD, the calprotectin assay reflects the activity could help identify novel markers in patients with familiar
of the disease, monitoring its progression, and can contribute history of EOCD. This could represent a major advance for
to the decision about the correct medication strategy. early-onset diagnosis as specific tests might be developed to
The application of this test in the pediatric population improve counselling, while direct identification of modifier
is a good result. Although the test is not yet able to replace genes might assist in the recognition of new genetic, environ-
diagnostic colonoscopy, it can be a good indicator for the mental, and microbial causes of CD.
decision to use or delay the use of invasive investigations [99].
Recently, Vitali et al. suggested the use of high-mobility Conflict of Interests
group box1 (HMGB1) as a novel marker of intestinal mucosal
inflammation. HMGB1 is today regarded as a pleiotropic The authors declare they have no conflict of interests related
cytokine, that is passively released by necrotic cells, but not to the issues discussed in this paper.
from apoptotic cells. Moreover HMGB1 could be actively
secreted from some types of immune cells in response
to lipopolysaccharide (LPS), IFN-𝛾, and TNF-𝛼. There is
Authors’ Contribution
evidence that HMGB1 is secreted in the stools of these Annalisa Marcuzzi and Anna Monica Bianco contributed
patients and not detectable in controls [101]. equally to this study.
The relationship between the genetic susceptibility and
the microbiome could be considered in the disease diagnosis.
There are several international human microbiome projects References
that have focused initially on the bacterial component of the [1] D. C. Baumgart and W. J. Sandborn, “Crohn’s disease,” The
microbiome. The evidence that bacteria play an important Lancet, vol. 380, no. 9853, pp. 1590–1605, 2012.
role in CD includes the observation that surgical diversion [2] G. Radford-Smith and N. Pandeya, “Associations between
of the fecal stream ameliorates the inflammation and that NOD2/CARD15 genotype and phenotype in Crohn’s disease—
antibiotics help some patients. Moreover other evidence is are we there yet?” World Journal of Gastroenterology, vol. 12, no.
showed in mouse models of colitis where virus, bacteria, 44, pp. 7097–7103, 2006.
or both acting together can contribute to the pathology via [3] D. K. Bonen and J. H. Cho, “The genetics of inflammatory bowel
signaling through innate immune sensors and regulation of disease,” Gastroenterology, vol. 124, no. 2, pp. 521–536, 2003.
pro- and anti-inflammatory cytokines [82]. The microbiome [4] S. K. Yang, S. Yun, J. H. Kim et al., “Epidemiology of
varies from person to person and such variation could pro- inflammatory bowel disease in the Songpa-Kangdong district,
vide environmental inputs that contribute to the incidence of Seoul, Korea, 1986–2005: a KASID study,” Inflammatory Bowel
CD, within the genetic foundation revealed by GWAS [102]. Diseases, vol. 14, no. 4, pp. 542–549, 2008.
The concept of dysbiosis as a contributor to CD is correlated [5] R. J. Xavier and D. K. Podolsky, “Unravelling the pathogenesis
with intestinal bacteria communities, so the changes in the of inflammatory bowel disease,” Nature, vol. 448, no. 7152, pp.
bacterial microbiota could have a potential role in the disease. 427–434, 2007.
However, this hypothesis needs to be expanded to include [6] J. Cosnes, S. Cattan, A. Blain et al., “Long-term evolution
specific interactions between individual bacteria and host of disease behavior of Crohn’s disease,” Inflammatory Bowel
genes. Diseases, vol. 8, no. 4, pp. 244–250, 2002.
Moreover, another ambitious goal is the identification [7] E. Louis, A. Collard, A. F. Oger, E. Degroote, F. A. Aboul Nasr
of a genetic pattern able to associate specific phenotypic El Yafi, and J. Belaiche, “Behaviour of Crohn’s disease according
to the Vienna classification: changing pattern over the course of
characteristics to CD patients or to anticipate the possible
the disease,” Gut, vol. 49, no. 6, pp. 777–782, 2001.
consequences of the disease already in childhood and thus
[8] L. Beaugerie, P. Seksik, I. Nion-Larmurier, J. P. Gendre, and J.
prevent complications associated with the disease and to
Cosnes, “Predictors of Crohn’s disease,” Gastroenterology, vol.
choose the best treatment for each patient. 130, no. 3, pp. 650–656, 2006.
A rapid diagnosis is fundamental to avoid a growth
[9] M. B. Heyman, B. S. Kirschner, B. D. Gold et al., “Children
delay or complications of the disease typical of the pediatric with early-onset inflammatory bowel disease (IBD): analysis of
disease leading to surgery. In some cases, genetic studies have a pediatric IBD consortium registry,” Journal of Pediatrics, vol.
provided useful information for the identification of specific 146, no. 1, pp. 35–40, 2005.
mutations that predict risk of stenosis and surgery and/or [10] Z. Cannioto, I. Berti, S. Martelossi et al., “IBD and IBD
disease localization in pediatric-onset CD [103, 104]. mimicking enterocolitis in children younger than 2 years of
The difficulty of finding a common genetic pattern of age,” European Journal of Pediatrics, vol. 168, no. 2, pp. 149–155,
association is caused by the multifactorial feature of the 2009.
disease that shows different characterizations by world region [11] IBD Working Group of the European Society for Paediatric
and race. Gastroenterology, Hepatology and Nutrition, “Inflammatory
BioMed Research International 9

bowel disease in children and adolescents: recommendations [29] L. D. Notarangelo, “Primary immunodeficiencies,” The Journal
for diagnosis—the Porto criteria,” Journal of Pediatric Gastroen- of Allergy and Clinical Immunology, vol. 125, no. 2, supplement
terology and Nutrition, vol. 41, no. 1, pp. 1–7, 2005. 2, pp. S182–S194, 2010.
[12] C. Spray, G. D. Debelle, and M. S. Murphy, “Current diagnosis, [30] A. Tommasini, A. Pirrone, G. Palla et al., “The universe of
management and morbidity in paediatric inflammatory bowel immune deficiencies in Crohn’s disease: a new viewpoint for an
disease,” Acta Paediatrica, vol. 90, no. 4, pp. 400–405, 2001. old disease?” Scandinavian Journal of Gastroenterology, vol. 45,
[13] J. D. Lewis, O. Abramson, M. Pascua et al., “Timing of no. 10, pp. 1141–1149, 2010.
myelosuppression during thiopurine therapy for inflammatory [31] L. de Ridder, R. K. Weersma, G. Dijkstra et al., “Genetic
bowel disease: implications for monitoring recommendations,” susceptibility has a more important role in pediatric-onset
Clinical Gastroenterology and Hepatology, vol. 7, no. 11, pp. 1195– Crohn’s disease than in adult-onset Crohn’s disease,” Inflamma-
1201, 2009. tory Bowel Diseases, vol. 13, no. 9, pp. 1083–1092, 2007.
[14] K. Mitsuyama, N. Tomiyasu, K. Takaki et al., “Interleukin-10 in [32] V. Biank, U. Broeckel, and S. Kugathasan, “Pediatric inflamma-
the pathophysiology of inflammatory bowel disease: increased tory bowel disease: clinical and molecular genetics,” Inflamma-
serum concentrations during the recovery phase,” Mediators of tory Bowel Diseases, vol. 13, no. 11, pp. 1430–1438, 2007.
Inflammation, vol. 2006, Article ID 26875, 7 pages, 2006. [33] M. Lacher, R. Kappler, S. Berkholz, H. Baurecht, D. von
[15] S. J. Bickston, L. W. Comerford, and F. Cominelli, “Future thera- Schweinitz, and S. Koletzko, “Association of a CXCL9 poly-
pies for inflammatory bowel disease,” Current Gastroenterology morphism with pediatric Crohn’s disease,” Biochemical and
Reports, vol. 5, no. 6, pp. 518–523, 2003. Biophysical Research Communications, vol. 363, no. 3, pp. 701–
[16] J. K. Hou, H. El-Serag, and S. Thirumurthi, “Distribution and 707, 2007.
manifestations of inflammatory bowel disease in Asians, His- [34] E. O. Glocker, D. Kotlarz, K. Boztug et al., “Inflammatory bowel
panics, and African Americans: a systematic review,” American disease and mutations affecting the interleukin-10 receptor,” The
Journal of Gastroenterology, vol. 104, no. 8, pp. 2100–2109, 2009. New England Journal of Medicine, vol. 361, no. 21, pp. 2033–2045,
[17] C. Abraham and J. H. Cho, “Inflammatory bowel disease,” The 2009.
New England Journal of Medicine, vol. 361, no. 21, pp. 2066– [35] M. Imielinski, R. N. Baldassano, A. Griffiths et al., “Common
2078, 2009. variants at five new loci associated with early-onset inflamma-
[18] N. Barnich, F. A. Carvalho, A. L. Glasser et al., “CEACAM6 tory bowel disease,” Nature Genetics, vol. 41, no. 12, pp. 1335–
acts as a receptor for adherent-invasive E. coli, supporting ileal 1340, 2009.
mucosa colonization in Crohn disease,” The Journal of Clinical
[36] D. K. Amre, D. R. Mack, K. Morgan et al., “Association between
Investigation, vol. 117, no. 6, pp. 1566–1574, 2007.
genome-wide association studies reported SNPs and pediatric-
[19] D. C. Baumgart and W. J. Sandborn, “Inflammatory bowel onset Crohn’s disease in Canadian children,” Human Genetics,
disease: clinical aspects and established and evolving therapies,” vol. 128, no. 2, pp. 131–135, 2010.
The Lancet, vol. 369, no. 9573, pp. 1641–1657, 2007.
[37] K. L. Helbig, M. Nothnagel, J. Hampe et al., “A case-only study
[20] D. K. Podolsky, “The current future understanding of inflam-
of gene-environment interaction between genetic susceptibility
matory bowel disease,” Best Practice & Research Clinical Gas-
variants in NOD2 and cigarette smoking in Crohn’s disease
troenterology, vol. 16, no. 6, pp. 933–943, 2002.
aetiology,” BMC Medical Genetics, vol. 13, article 14, 2012.
[21] A. Gutierrez, M. Scharl, L. Sempere et al., “Genetic susceptibility
[38] C. Kunz, S. Kuntz, and S. Rudloff, “Intestinal flora,” Advances in
to increased bacterial translocation influences the response to
Experimental Medicine and Biology, vol. 639, pp. 67–79, 2009.
biological therapy in patients with Crohn’s disease,” Gut, 2013.
[22] J. P. Achkar and R. Duerr, “The expanding universe of inflam- [39] W. B. Whitman, D. C. Coleman, and W. J. Wiebe, “Prokaryotes:
matory bowel disease genetics,” Current Opinion in Gastroen- the unseen majority,” Proceedings of the National Academy of
terology, vol. 24, no. 4, pp. 429–434, 2008. Sciences of the United States of America, vol. 95, no. 12, pp. 6578–
6583, 1998.
[23] W. Bodmer and I. Tomlinson, “Rare genetic variants and the
risk of cancer,” Current Opinion in Genetics & Development, vol. [40] R. E. Ley, D. A. Peterson, and J. I. Gordon, “Ecological and
20, no. 3, pp. 262–267, 2010. evolutionary forces shaping microbial diversity in the human
intestine,” Cell, vol. 124, no. 4, pp. 837–848, 2006.
[24] D. A. van Heel, S. Ghosh, M. Butler et al., “Muramyl dipeptide
and toll-like receptor sensitivity in NOD2-associated Crohn’s [41] H. Verstraelen, “Cutting edge: the vaginal microflora and bac-
disease,” The Lancet, vol. 365, no. 9473, pp. 1794–1796, 2005. terial vaginosis,” Verhandelingen—Koninklijke Academie voor
[25] J. van Limbergen, D. C. Wilson, and J. Satsangi, “The genetics Geneeskunde van België, vol. 70, no. 3, pp. 147–174, 2008.
of Crohn’s disease,” Annual Review of Genomics and Human [42] J. I. Gordon, “Honor thy gut symbionts redux,” Science, vol. 336,
Genetics, vol. 10, pp. 89–116, 2009. no. 6086, pp. 1251–1253, 2012.
[26] C. W. Lees, J. C. Barrett, M. Parkes, and J. Satsangi, “New IBD [43] D. N. Frank, A. L. St Amand, R. A. Feldman, E. C. Boedeker,
genetics: common pathways with other diseases,” Gut, vol. 60, N. Harpaz, and N. R. Pace, “Molecular-phylogenetic character-
no. 12, pp. 1739–1753, 2011. ization of microbial community imbalances in human inflam-
[27] S. Haglund, S. Almer, C. Peterson, and J. Söderman, “Gene matory bowel diseases,” Proceedings of the National Academy
expression and thiopurine metabolite profiling in inflammatory of Sciences of the United States of America, vol. 104, no. 34, pp.
bowel disease—novel clues to drug targets and disease mecha- 13780–13785, 2007.
nisms?” PLoS ONE, vol. 8, no. 2, Article ID e56989, 2013. [44] V. Redondo-Lopez, R. L. Cook, and J. D. Sobel, “Emerging role
[28] L. A. Hindorff, P. Sethupathy, H. A. Junkins et al., “Potential eti- of lactobacilli in the control and maintenance of the vaginal
ologic and functional implications of genome-wide association bacterial microflora,” Reviews of Infectious Diseases, vol. 12, no.
loci for human diseases and traits,” Proceedings of the National 5, pp. 856–872, 1990.
Academy of Sciences of the United States of America, vol. 106, no. [45] A. Huurre, M. Kalliomäki, S. Rautava, M. Rinne, S. Salminen,
23, pp. 9362–9367, 2009. and E. Isolauri, “Mode of delivery—effects on gut microbiota
10 BioMed Research International

and humoral immunity,” Neonatology, vol. 93, no. 4, pp. 236– [62] J. P. Y. Ting, D. L. Kastner, and H. M. Hoffman, “CATER-
240, 2008. PILLERs, pyrin and hereditary immunological disorders,”
[46] E. G. Zoetendal, A. von Wright, T. Vilpponen-Salmela, K. Ben- Nature Reviews Immunology, vol. 6, no. 3, pp. 183–195, 2006.
Amor, A. D. L. Akkermans, and W. M. de Vos, “Mucosa- [63] S. Brand, “Crohn’s disease: Th1, Th17 or both? The change of a
associated bacteria in the human gastrointestinal tract are paradigm: new immunological and genetic insights implicate
uniformly distributed along the colon and differ from the Th17 cells in the pathogenesis of Crohn’s disease,” Gut, vol. 58,
community recovered from feces,” Applied and Environmental no. 8, pp. 1152–1167, 2009.
Microbiology, vol. 68, no. 7, pp. 3401–3407, 2002. [64] T. S. Plantinga, L. A. Joosten, J. W. van der Meer, and M.
[47] R. E. Ley, F. Bäckhed, P. Turnbaugh, C. A. Lozupone, R. D. G. Netea, “Modulation of inflammation by autophagy: conse-
Knight, and J. I. Gordon, “Obesity alters gut microbial ecology,” quences for Crohn’s disease,” Current Opinion in Pharmacology,
Proceedings of the National Academy of Sciences of the United vol. 12, no. 4, pp. 497–502, 2012.
States of America, vol. 102, no. 31, pp. 11070–11075, 2005. [65] B. Levine and V. Deretic, “Unveiling the roles of autophagy in
[48] C. Zhang, M. Zhang, S. Wang et al., “Interactions between gut innate and adaptive immunity,” Nature Reviews Immunology,
microbiota, host genetics and diet relevant to development of vol. 7, no. 10, pp. 767–777, 2007.
metabolic syndromes in mice,” The ISME Journal, vol. 4, no. 2, [66] V. Deretic, “Autophagy as an innate immunity paradigm:
pp. 232–241, 2010. expanding the scope and repertoire of pattern recognition
[49] S. Rakoff-Nahoum and R. Medzhitov, “Innate immune recog- receptors,” Current Opinion in Immunology, vol. 24, no. 1, pp.
nition of the indigenous microbial flora,” Mucosal Immunology, 21–31, 2012.
vol. 1, supplement 1, pp. S10–S14, 2008. [67] A. C. Villani, M. Lemire, G. Fortin et al., “Common variants in
[50] P. B. Eckburg, E. M. Bik, C. N. Bernstein et al., “Diversity of the the NLRP3 region contribute to Crohn’s disease susceptibility,”
human intestinal microbial flora,” Science, vol. 308, no. 5728, pp. Nature Genetics, vol. 41, no. 1, pp. 71–76, 2009.
1635–1638, 2005. [68] L. Ferrero-Miliani, O. H. Nielsen, P. S. Andersen, and S. E.
[51] M. Baumgart, B. Dogan, M. Rishniw et al., “Culture indepen- Girardin, “Chronic inflammation: importance of NOD2 and
dent analysis of ileal mucosa reveals a selective increase in NALP3 in interleukin-1𝛽 generation,” Clinical and Experimental
invasive Escherichia coli of novel phylogeny relative to depletion Immunology, vol. 147, no. 2, pp. 227–235, 2007.
of Clostridiales in Crohn’s disease involving the ileum,” The [69] A. Gombault, L. Baron, and I. Couillin, “ATP release and
ISME Journal, vol. 1, no. 5, pp. 403–418, 2007. purinergic signaling in NLRP3 inflammasome activation,” Fron-
[52] E. Li, C. M. Hamm, A. S. Gulati et al., “Inflammatory bowel dis- tiers in Immunology, vol. 3, article 414, 2012.
eases phenotype, C. difficile and NOD2 genotype are associated [70] T. D. Kanneganti, N. Özören, M. Body-Malapel et al., “Bacterial
with shifts in human ileum associated microbial composition,” RNA and small antiviral compounds activate caspase-1 through
PLoS ONE, vol. 7, no. 6, Article ID e26284, 2012. cryopyrin/NALP3,” Nature, vol. 440, no. 7081, pp. 233–236,
[53] H. Sokol and P. Seksik, “The intestinal microbiota in inflam- 2006.
matory bowel diseases: time to connect with the host,” Current [71] S. R. Kingsbury, P. G. Conaghan, and M. F. McDermott,
Opinion in Gastroenterology, vol. 26, no. 4, pp. 327–331, 2010. “The role of the NLRP3 inflammasome in gout,” Journal of
[54] M. Joossens, G. Huys, M. Cnockaert et al., “Dysbiosis of the Inflammation Research, vol. 4, no. 1, pp. 39–49, 2011.
faecal microbiota in patients with Crohn’s disease and their [72] F. Martinon, L. Agostini, E. Meylan, and J. Tschopp, “Iden-
unaffected relatives,” Gut, vol. 60, no. 5, pp. 631–637, 2011. tification of bacterial muramyl dipeptide as activator of the
[55] H. W. Virgin, E. J. Wherry, and R. Ahmed, “Redefining chronic NALP3/cryopyrin inflammasome,” Current Biology, vol. 14, no.
viral infection,” Cell, vol. 138, no. 1, pp. 30–50, 2009. 21, pp. 1929–1934, 2004.
[56] E. Elinav, J. Henao-Mejia, and R. A. Flavell, “Integrative inflam- [73] S. Mariathasan and D. M. Monack, “Inflammasome adaptors
masome activity in the regulation of intestinal mucosal immune and sensors: intracellular regulators of infection and inflamma-
responses,” Mucosal Immunology, vol. 6, no. 1, pp. 4–13, 2013. tion,” Nature Reviews Immunology, vol. 7, no. 1, pp. 31–40, 2007.
[57] D. N. Frank, C. E. Robertson, C. M. Hamm et al., “Disease [74] E. Elinav, T. Strowig, A. L. Kau et al., “NLRP6 inflammasome
phenotype and genotype are associated with shifts in intestinal- regulates colonic microbial ecology and risk for colitis,” Cell, vol.
associated microbiota in inflammatory bowel diseases,” Inflam- 145, no. 5, pp. 745–757, 2011.
matory Bowel Diseases, vol. 17, no. 1, pp. 179–184, 2011. [75] O. von Kampen, S. Lipinski, A. Till et al., “Caspase recruitment
[58] U. Gophna, K. Sommerfeld, S. Gophna, W. F. Doolittle, and domain-containing protein 8 (CARD8) negatively regulates
S. J. O. Veldhuyzen van Zanten, “Differences between tissue- NOD2-mediated signaling,” The Journal of Biological Chemistry,
associated intestinal microfloras of patients with Crohn’s disease vol. 285, no. 26, pp. 19921–19926, 2010.
and ulcerative colitis,” Journal of Clinical Microbiology, vol. 44, [76] J. Hampe, A. Franke, P. Rosenstiel et al., “A genome-wide asso-
no. 11, pp. 4136–4141, 2006. ciation scan of nonsynonymous SNPs identifies a susceptibility
[59] C. Manichanh, L. Rigottier-Gois, E. Bonnaud et al., “Reduced variant for Crohn disease in ATG16L1,” Nature Genetics, vol. 39,
diversity of faecal microbiota in Crohn’s disease revealed by a no. 2, pp. 207–211, 2007.
metagenomic approach,” Gut, vol. 55, no. 2, pp. 205–211, 2006. [77] S. A. McCarroll, A. Huett, P. Kuballa et al., “Deletion poly-
[60] K. Cadwell, “Crohn’s disease susceptibility gene interactions, a morphism upstream of IRGM associated with altered IRGM
NOD to the newcomer ATG16L1,” Gastroenterology, vol. 139, no. expression and Crohn’s disease,” Nature Genetics, vol. 40, no. 9,
5, pp. 1448–1450, 2010. pp. 1107–1112, 2008.
[61] D. M. Glubb, R. B. Gearry, M. L. Barclay et al., “NOD2 and [78] R. Cooney, J. Baker, O. Brain et al., “NOD2 stimulation induces
ATG16L1 polymorphisms affect monocyte responses in Crohn’s autophagy in dendritic cells influencing bacterial handling and
disease,” World Journal of Gastroenterology, vol. 17, no. 23, pp. antigen presentation,” Nature Medicine, vol. 16, no. 1, pp. 90–97,
2829–2837, 2011. 2010.
BioMed Research International 11

[79] L. H. Travassos, L. A. M. Carneiro, M. Ramjeet et al., “Nod1 and [95] A. Darfeuille-Michaud, C. Neut, N. Barnich et al., “Presence of
NOD2 direct autophagy by recruiting ATG16L1 to the plasma adherent Escherichia coli strains in ileal mucosa of patients with
membrane at the site of bacterial entry,” Nature Immunology, Crohn’s disease,” Gastroenterology, vol. 115, no. 6, pp. 1405–1413,
vol. 11, no. 1, pp. 55–62, 2010. 1998.
[80] D. Glick, S. Barth, and K. F. Macleod, “Autophagy: cellular and [96] S. B. Singh, W. Ornatowski, I. Vergne et al., “Human IRGM
molecular mechanisms,” The Journal of Pathology, vol. 221, no. regulates autophagy and cell-autonomous immunity functions
1, pp. 3–12, 2010. through mitochondria,” Nature Cell Biology, vol. 12, no. 12, pp.
[81] A. Huett and R. J. Xavier, “Autophagy at the gut interface: 1154–1165, 2010.
mucosal responses to stress and the consequences for inflam- [97] I. P. Gregoire, C. Richetta, L. Meyniel-Schicklin et al., “IRGM
matory bowel diseases,” Inflammatory Bowel Diseases, vol. 16, is a common target of RNA viruses that subvert the autophagy
no. 1, pp. 152–174, 2010. network,” PLoS Pathogens, vol. 7, no. 12, Article ID e1002422,
[82] J. M. Yuk, T. Yoshimori, and E. K. Jo, “Autophagy and bacterial 2011.
infectious diseases,” Experimental & Molecular Medicine, vol. [98] E. Papa, M. Docktor, C. Smillie et al., “Non-invasive mapping of
44, no. 2, pp. 99–108, 2012. the gastrointestinal microbiota identifies children with inflam-
[83] C. R. Homer, A. L. Richmond, N. A. Rebert, J. Achkar, and matory bowel disease,” PLoS ONE, vol. 7, no. 6, Article ID
C. McDonald, “ATG16L1 and NOD2 interact in an autophagy- e39242,, 2012.
dependent antibacterial pathway implicated in Crohn’s disease [99] R. M. Ayling, “New faecal tests in gastroenterology,” Annals of
pathogenesis,” Gastroenterology, vol. 139, no. 5, pp. 1630.e2– Clinical Biochemistry, vol. 49, part 1, pp. 44–54, 2012.
1641.e2, 2010. [100] P. F. van Rheenen, E. van de Vijver, and V. Fidler, “Faecal
[84] B. C. Miller, Z. Zhao, L. M. Stephenson et al., “The autophagy calprotectin for screening of patients with suspected inflamma-
gene ATG5 plays an essential role in B lymphocyte develop- tory bowel disease: diagnostic meta-analysis,” British Medical
ment,” Autophagy, vol. 4, no. 3, pp. 309–314, 2008. Journal, vol. 341, p. c3369, 2010.
[85] P. Rosenstiel, G. Jacobs, A. Till, and S. Schreiber, “NOD-like [101] R. Vitali, L. Stronati, A. Negroni et al., “Fecal HMGB1 is a
receptors: ancient sentinels of the innate immune system,” novel marker of intestinal mucosal inflammation in pediatric
Cellular and Molecular Life Sciences, vol. 65, no. 9, pp. 1361–1377, inflammatory bowel disease,” The American Journal of Gas-
2008. troenterology, vol. 106, no. 11, pp. 2029–2040, 2011.
[86] S. J. Ott, M. Musfeldt, D. F. Wenderoth et al., “Reduction in [102] S. Ehlers, S. H. E. Kaufmann, and Participants of the 99th
diversity of the colonic mucosa associated bacterial microflora Dahlem Conference, “Infection, inflammation, and chronic dis-
in patients with active inflammatory bowel disease,” Gut, vol. 53, eases: consequences of a modern lifestyle,” Trends in Immunol-
no. 5, pp. 685–693, 2004. ogy, vol. 31, no. 5, pp. 184–190, 2010.
[87] J. Qin, R. Li, J. Raes et al., “A human gut microbial gene catalogue [103] A. Marcuzzi, M. Girardelli, A. M. Bianco et al., “Inflammation
established by metagenomic sequencing,” Nature, vol. 464, no. profile of four early onset Crohn patients,” Gene, vol. 493, no. 2,
7285, pp. 59–65, 2010. pp. 282–285, 2012.
[88] Z. M. Kanaan, M. R. Eichenberger, S. Ahmad et al., “Clinical [104] A. M. Bianco, V. Zanin, M. Girardelli et al., “A common genetic
predictors of inflammatory bowel disease in a genetically well- background could explain early-onset Crohn’s disease,” Medical
defined Caucasian population,” Journal of Negative Results in Hyphoteses, vol. 78, no. 4, pp. 520–522, 2012.
Biomedicine, vol. 11, article 7, 2012.
[89] S. Lesage, H. Zouali, J. P. Cezard et al., “CARD15/NOD2
mutational analysis and genotype-phenotype correlation in
612 patients with inflammatory bowel disease,” The American
Journal of Human Genetics, vol. 70, no. 4, pp. 845–857, 2002.
[90] T. Petnicki-Ocwieja, T. Hrncir, Y. J. Liu et al., “NOD2 is required
for the regulation of commensal microbiota in the intestine,”
Proceedings of the National Academy of Sciences of the United
States of America, vol. 106, no. 37, pp. 15813–15818, 2009.
[91] P. Kuballa, A. Huett, J. D. Rioux, M. J. Daly, and R. J. Xavier,
“Impaired autophagy of an intracellular pathogen induced by a
Crohn’s disease associated ATG16L1 variant,” PLoS ONE, vol. 3,
no. 10, Article ID e3391, 2008.
[92] Y. G. Kim, N. Kamada, M. H. Shaw et al., “The NOD2 sensor
promotes intestinal pathogen eradication via the chemokine
CCL2-dependent recruitment of inflammatory monocytes,”
Immunity, vol. 34, no. 5, pp. 769–780, 2011.
[93] M. Parkes, J. C. Barrett, N. J. Prescott et al., “Sequence variants
in the autophagy gene IRGM and multiple other replicating loci
contribute to Crohn’s disease susceptibility,” Nature Genetics,
vol. 39, no. 7, pp. 830–832, 2007.
[94] P. Brest, P. Lapaquette, M. Souidi et al., “A synonymous variant
in IRGM alters a binding site for miR-196 and causes deregula-
tion of IRGM-dependent xenophagy in Crohn’s disease,” Nature
Genetics, vol. 43, no. 3, pp. 242–245, 2011.

Das könnte Ihnen auch gefallen